DOI QR코드

DOI QR Code

A formulated red ginseng extract inhibits autophagic flux and sensitizes to doxorubicin-induced cell death

  • Park, Han-Hee (Department of Biochemistry, Ajou University School of Medicine) ;
  • Choi, Seung-Won (Department of Biochemistry, Ajou University School of Medicine) ;
  • Lee, Gwang Jin (College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University) ;
  • Kim, Young-Dae (Department of Biochemistry, Ajou University School of Medicine) ;
  • Noh, Hyun-Jin (Department of Biochemistry, Ajou University School of Medicine) ;
  • Oh, Seung-Jae (Department of Biochemistry, Ajou University School of Medicine) ;
  • Yoo, Iseul (Department of Biochemistry, Ajou University School of Medicine) ;
  • Ha, Yu-Jin (Department of Biochemistry, Ajou University School of Medicine) ;
  • Koo, Gi-Bang (Department of Biochemistry, Ajou University School of Medicine) ;
  • Hong, Soon-Sun (College of Medicine, Inha University) ;
  • Kwon, Sung Won (College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University) ;
  • Kim, You-Sun (Department of Biochemistry, Ajou University School of Medicine)
  • Received : 2017.07.02
  • Accepted : 2017.08.14
  • Published : 2019.01.15

Abstract

Background: Ginseng is believed to have antitumor activity. Autophagy is largely a prosurvival cellular process that is activated in response to cellular stressors, including cytotoxic chemotherapy; therefore, agents that inhibit autophagy can be used as chemosensitizers in cancer treatment. We examined the ability of Korean Red Ginseng extract (RGE) to prevent autophagic flux and to make hepatocellular carcinoma (HCC) cells become more sensitive to doxorubicin. Methods: The cytotoxic effects of total RGE or its saponin fraction (RGS) on HCC cells were examined by the lactate dehydrogenase assay in a dose- or time-dependent manner. The effect of RGE or RGS on autophagy was measured by analyzing microtubule-associated protein 1A/1B-light chain (LC)3-II expression and LC3 puncta formation in HCC cells. Late-stage autophagy suppression was tested using tandem-labeled green fluorescent protein (GFP)-monomeric red fluorescent protein (mRFP)-LC3. Results: RGE markedly increased the amount of LC3-II, but green and red puncta in tandem-labeled GFP-mRFP-LC3 remained colocalized over time, indicating that RGE inhibited autophagy at a late stage. Suppression of autophagy through knockdown of key ATG genes increased doxorubicin-induced cell death, suggesting that autophagy induced by doxorubicin has a protective function in HCC. Finally, RGE and RGS markedly sensitized HCC cells, (but not normal liver cells), to doxorubicin-induced cell death. Conclusion: Our data suggest that inhibition of late-stage autophagic flux by RGE is important for its potentiation of doxorubicin-induced cancer cell death. Therapy combining RGE with doxorubicin could serve as an effective strategy in the treatment of HCC.

Keywords

References

  1. Jia L, Zhao Y, Liang XJ. Current evaluation of the millennium phytomedicineginseng (II): collected chemical entities, modern pharmacology, and clinical applications emanated from traditional Chinese medicine. Curr Med Chem 2009;16:2924-42. https://doi.org/10.2174/092986709788803204
  2. Seo EY, Kim WK. Red ginseng extract reduced metastasis of colon cancer cells in vitro and in vivo. J Ginseng Res 2011;35:315-24. https://doi.org/10.5142/jgr.2011.35.3.315
  3. Chen S, Wang Z, Huang Y, O'Barr SA, Wong RA, Yeung S, Chow MS. Ginseng and anticancer drug combination to improve cancer chemotherapy: a critical review. Evid Based Complement Alternat Med 2014;2014:1-13.
  4. Liu TG, Huang Y, Cui DD, Huang XB, Mao SH, Ji LL, Song HB, Yi C. Inhibitory effect of ginsenoside Rg3 combined with gemcitabine on angiogenesis and growth of lung cancer in mice. BMC Cancer 2009;9:250. https://doi.org/10.1186/1471-2407-9-250
  5. Zhang QY, Kang XM, Zhao WH. Antiangiogenic effect of low-dose cyclophosphamide combined with ginsenoside Rg3 on Lewis lung carcinoma. Biochem Biophys Res Comm 2006;342:824-8. https://doi.org/10.1016/j.bbrc.2006.02.044
  6. Keum YS, Han SS, Chun KS, Park KK, Park JH, Lee SK, Surh YJ. Inhibitory effects of the ginsenoside Rg3 on phorbol ester-induced cyclooxygenase-2 expression, NF-kappaB activation and tumor promotion. Mutat Res 2003;523-524:75-85. https://doi.org/10.1016/S0027-5107(02)00323-8
  7. Chen QJ, Zhang MZ, Wang LX. Gensenoside Rg3 inhibits hypoxia-induced VEGF expression in human cancer cells. Cell Physiol Biochem 2010;26:849-58. https://doi.org/10.1159/000323994
  8. Kim JW, Jung SY, Kwon YH, Lee JH, Lee YM, Lee BY, Kwon SM. Ginsenoside Rg3 attenuates tumor angiogenesis via inhibiting bioactivities of endothelial progenitor cells. Cancer Biol Ther 2012;13:504-15. https://doi.org/10.4161/cbt.19599
  9. Xu TM, Xin Y, Cui MH, Jiang X, Gu LP. Inhibitory effect of ginsenoside Rg3 combined with cyclophosphamide on growth and angiogenesis of ovarian cancer. Chinese Med J-Peking 2007;120:584-8. https://doi.org/10.1097/00029330-200704010-00011
  10. Lee YS, Lee DG, Lee JY, Kim TR, Hong SS, Kwon Kim YS. A formulated red ginseng extract upregulates CHOP and increases TRAIL-mediated cytotoxicity in human hepatocellular carcinoma cells. Int J Oncol 2013;43:591-9. https://doi.org/10.3892/ijo.2013.1964
  11. Lee JY, Jung KH, Morgan MJ, Kang YR, Lee HS, Koo GB, Hong SS, Kwon SW, Kim YS. Sensitization of TRAIL-induced cell death by 20(S)-ginsenoside Rg3 via CHOP-mediated DR5 upregulation in human hepatocellular carcinoma cells. Mol Cancer Ther 2013;12:274-85. https://doi.org/10.1158/1535-7163.MCT-12-0054
  12. White E. The role for autophagy in cancer. J Clin Invest 2015;125:42-6. https://doi.org/10.1172/JCI73941
  13. Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol 2007;8:741-52. https://doi.org/10.1038/nrm2239
  14. Shen HM, Codogno P. Autophagic cell death: Loch Ness monster or endangered species? Autophagy 2011;7:457-65. https://doi.org/10.4161/auto.7.5.14226
  15. Rosenfeldt Mathias T, Ryan Kevin M. The multiple roles of autophagy in cancer. Carcinogenesis 2011;32:955. https://doi.org/10.1093/carcin/bgr031
  16. Mazzanti R, Arena U, Tassi R. Hepatocellualr carcinoma: where are we? World J Exp Med 2016;20:21-36. https://doi.org/10.5493/wjem.v6.i1.21
  17. Takimoto CH, Awada A. Safety and anti-tumor activity of sorafenib (Nexavar) in combination with other anti-cancer agents: a review of clinical trials. Cancer Chemother Pharmacol 2008;61:535-48. https://doi.org/10.1007/s00280-007-0639-9
  18. Cabibbo G, Latteri F, Antonucci M, Craxi A. Multimodal approaches to the treatment of hepatocellular carcinoma. Nat Clin Pract Gastroenterol Hepatol 2009;6:159-69. https://doi.org/10.1038/ncpgasthep1357
  19. Shi YH, Ding ZB, Zhou J, Hui B, Shi GM, Ke AW, Wang XY, Dai Z, Peng YF, Gu CY, et al. Targeting autophagy enhances sorafenib lethality for hepatocellular carcinoma via ER stress-related apoptosis. Autophagy 2011;7:1159-72. https://doi.org/10.4161/auto.7.10.16818
  20. Ding ZB, Hui B, Shi YH, Zhou J, Peng YF, Gu CY, Yang H, Shi GM, Ke AW, Wang XY, et al. Autophagy activation in hepatocellular carcinoma contributes to the tolerance of oxaliplatin via reactive oxygen species modulation. Clin Cancer Res 2011;17:6229-38. https://doi.org/10.1158/1078-0432.CCR-11-0816
  21. Kim DG, Jung KH, Lee DG, Yoon JH, Choi KS, Kwon SW, Shen HM, Morgan MJ, Hong SS, Kim YS. 20(S)-Ginsenoside Rg3 is a novel inhibitor of autophagy and sensitizeshepatocellular carcinoma todoxorubicin.Oncotarget2014;5:4438-51. https://doi.org/10.18632/oncotarget.2034
  22. Semenov D, Lushnikova E, Nepomnyashchikh L. Anthracycline-induced cardiomyopathy is manifested in decreased protein synthesis, impaired intracellular regeneration, and non-necrotic death of cardiomyocytes. Bull Exp Biol Med 2001;131:505-10. https://doi.org/10.1023/A:1017956922385
  23. Park JY, Choi P, Lee D, Kim T, Jung EB, Hwang BS, Kang KS, Ham J. Effect of amino acids on the generation of ginsenoside Rg3 epimers by heat processing and the anticancer activities of epimers in A2780 human ovarian cancer cells. Evid Based Complement Alternat Med 2016;20:1-6.
  24. Kaur J, Debnath J. Autophagy at the crossroads of catabolism and anabolism. Nat Rev Mol Cell Biol 2015;16:461-72. https://doi.org/10.1038/nrm4024
  25. Pankiv S, Clausen T, Lamark T, Brech A, Bruun J, Outzen H, Overvatn A, Bjorkoy G, Johansen T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 2007;282:24131-45. https://doi.org/10.1074/jbc.M702824200
  26. Mizushima N, Yoshimori T. How to interpret LC3 immunoblotting. Autophagy 2007;3:542-5. https://doi.org/10.4161/auto.4600
  27. Bjorkoy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, Stenmark H, Johansen T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 2005;171:603-14. https://doi.org/10.1083/jcb.200507002
  28. Kimura S, Noda T, Yoshimori T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 2007;3:452-60. https://doi.org/10.4161/auto.4451
  29. Katayama H, Yamamoto A, Mizushima N, Yoshimori T, Miyawaki A. GFP-like proteins stably accumulate in lysosomes. Cell Struct Funct 2007;33:1-12. https://doi.org/10.1247/csf.07011
  30. Yang ZJ, Chee CE, Huang S, Sinicrope FA. The role of autophagy in cancer: therapeutic implications. Mol Cancer Ther 2011;10:1533-41. https://doi.org/10.1158/1535-7163.MCT-11-0047
  31. Chang MS, Lee SG, Rho HM. Transcriptional activation of Cu/Zn superoxide dismutase and catalase genes by panaxadiol ginsenosides extracted from Panax ginseng. Phytother Res 1999;13:641-4. https://doi.org/10.1002/(SICI)1099-1573(199912)13:8<641::AID-PTR527>3.0.CO;2-Z
  32. Sui X, Chen R, Wang Z, Huang Z, Kong N, Zhang M, Han W, Lou F, Yang J, Zhang Q, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 2013;10:e838.
  33. Morgan MJ, Gamez G, Menke C, Hernandez A, Thorburn J, Gidan F, Staskiewicz L, Morgan S, Cummings C, Maycotte P, et al. Regulation of autophagy and chloroquine sensitivity by oncogenic RAS in vitro is contextdependent. Autophagy 2014;10:1814-26. https://doi.org/10.4161/auto.32135
  34. Xu XD, Zhao Y, Zhang M, He RZ, Shi XH, Guo XJ, Shi CJ, Peng F, Wang M, Shen M, et al. Inhibition of autophagy by deguelin sensitizes pancreatic cancer cells to doxorubicin. Int J Mol Sci 2017;18:1-13. https://doi.org/10.3390/ijms18010001

Cited by

  1. Concomitant Inhibition of Cytoprotective Autophagy Augments the Efficacy of Withaferin A in Hepatocellular Carcinoma vol.11, pp.4, 2019, https://doi.org/10.3390/cancers11040453
  2. hERG1 is involved in the pathophysiological process and inhibited by berberine in SKOV3 cells vol.17, pp.6, 2019, https://doi.org/10.3892/ol.2019.10263
  3. The Role of Autophagy in Liver Cancer: Crosstalk in Signaling Pathways and Potential Therapeutic Targets vol.13, pp.12, 2019, https://doi.org/10.3390/ph13120432
  4. Autophagy, an accomplice or antagonist of drug resistance in HCC? vol.12, pp.3, 2021, https://doi.org/10.1038/s41419-021-03553-7
  5. Targeting autophagy using saponins as a therapeutic and preventive strategy against human diseases vol.166, 2019, https://doi.org/10.1016/j.phrs.2021.105428